ArticlePDF Available

Voltage-dependent anion channel localises to the plasma membrane and peripheral but not perinuclear mitochondria

Authors:

Abstract and Figures

Activity of the antioestrogen-activated maxi-Cl(-) channel has been recorded in different cell types, including fibroblasts, vascular smooth muscle, endothelial and neuroblastoma cells. Its electrophysiological properties resemble those of the voltage-dependent anion channel (VDAC) of the outer mitochondrial membrane, a channel of particular relevance to the physiology and pathophysiology of mitochondria. The hypothesis that VDAC could be the molecular correlate of the plasma membrane maxi-Cl(-) channel has been debated over the last few years, with the lack of clear evidence for the presence of VDAC in the plasma membrane constituting the main argument of the detractors. In the present study, we investigated the cellular localisation of VDAC in NIH3T3 fibroblasts. The presence of a plasma membrane VDAC was demonstrated by immunoblotting of membrane fractions with monoclonal antibodies against the VDAC and by RT-PCR using primers that hybridise to a VDAC sequence coding for a N-terminal leader peptide required for its plasma membrane sorting. In addition, confocal microscopy studies showed the colocalisation of VDAC with caveolin-1. As expected, VDAC also localised to mitochondria. Colocalisation studies with TOM-20, a protein also present in the outer mitochondrial membrane, showed that VDAC proteins localised only to peripheral and not to perinuclear mitochondria.
Content may be subject to copyright.
Pflugers Arch - Eur J Physiol (2003) 446:309–313
DOI 10.1007/s00424-003-1054-7
EJP (PFLGERS ARCHIV)—SYMPOSIUM
Maria I. Bahamonde · Miguel A. Valverde
Voltage-dependent anion channel localises to the plasma membrane
and peripheral but not perinuclear mitochondria
Published online: 16 April 2003
Springer-Verlag 2003
Abstract Activity of the antioestrogen-activated maxi-
Clchannel has been recorded in different cell types,
including fibroblasts, vascular smooth muscle, endothelial
and neuroblastoma cells. Its electrophysiological proper-
ties resemble those of the voltage-dependent anion
channel (VDAC) of the outer mitochondrial membrane,
a channel of particular relevance to the physiology and
pathophysiology of mitochondria. The hypothesis that
VDAC could be the molecular correlate of the plasma
membrane maxi-Clchannel has been debated over the
last few years, with the lack of clear evidence for the
presence of VDAC in the plasma membrane constituting
the main argument of the detractors. In the present study,
we investigated the cellular localisation of VDAC in
NIH3T3 fibroblasts. The presence of a plasma membrane
VDAC was demonstrated by immunoblotting of mem-
brane fractions with monoclonal antibodies against the
VDAC and by RT-PCR using primers that hybridise to a
VDAC sequence coding for a N-terminal leader peptide
required for its plasma membrane sorting. In addition,
confocal microscopy studies showed the colocalisation of
VDAC with caveolin-1. As expected, VDAC also local-
ised to mitochondria. Colocalisation studies with TOM-
20, a protein also present in the outer mitochondrial
membrane, showed that VDAC proteins localised only to
peripheral and not to perinuclear mitochondria.
Keywords VDAC · Maxi-Clchannel · Lipid rafts ·
Caveolin · Antioestrogen · Mitochondria · Fibroblasts ·
Neuroblastoma
Introduction
Voltage-dependent anion channels (VDACs), also known
as porins, are integral membrane proteins which form a
pore slightly more permeable to anions than cations, and
are also permeable to small solutes [1]. They have been
identified in the mitochondrial outer membrane where
they provide a major pathway for the transport of
metabolites, e.g. ATP [2] and cholesterol [3], among
others. They are also involved in the mitochondrial events
leading to apoptosis [4, 5]. Besides their mitochondrial
location, several reports claim the presence of VDAC in
the plasma membrane of different cell types [6, 7, 8, 9].
The fact that its electrophysiological properties [1]
resemble those of the plasma membrane maxi-Clchan-
nel [10, 11, 12] has encouraged some investigators to
assume that the two channels were one and the same
protein [6, 7, 8, 9, 10, 11, 12, 13]. This hypothesis was
based on early observations suggesting the presence of
VDAC protein on the plasma membrane [7, 8, 9, 10, 11,
12, 13] but has been questioned by others [14]. The extra-
mitochondrial location of VDAC has recently received
strong support from two independent reports: the identi-
fication of a VDAC isoform (pl-VDAC) that contains a
leader sequence for its trafficking to the plasma mem-
brane [9] and the presence of VDAC in caveolae [8].
In the present study, in addition to demonstrating the
plasma membrane location of VDAC and its association
with caveolae in NIH3T3 fibroblasts, we provide the first
evidence suggesting its preferential localisation to a
peripheral pool of mitochondria.
Materials and methods
Cell culture
NIH3T3 mouse fibroblasts permanently transfected with the MDR1
gene were cultured as previously described [11]. When confluent,
cells were either subcultured into T-75 flasks (Nunc) and used
either for immunoblotting or plated on to treated glass cover-slips
for immunofluorescence experiments.
M. I. Bahamonde · M. A. Valverde ())
Unitat de Senyalitzaci Cel·lular,
Departament de Cincies Experimentals i de la Salut,
Universitat Pompeu Fabra, C/Dr. Aiguader 80,
08003 Barcelona, Spain
e-mail: miguel.valverde@cexs.upf.es
Fax: +34-93-5422802
Immunoblotting
Plasma membrane proteins and cell lysate were prepared using a
previously described protocol [15, 16]. Proteins were resolved by
SDS-PAGE (12%) and blotted onto nitrocellulose. The primary
antibody used was the monoclonal mouse anti-porin 31 HL
(VDAC1) (Ab-2 antibody, Calbiochem; 1:2,000 dilution). Non-
specific binding was avoided by incubating the nitrocellulose
membranes in a blocking solution consisting of TTBS buffer
(100 mM TRIS-HCl pH 7.5, 150 mM NaCl) supplemented with 5%
non-fat milk for 1 h at room temperature or overnight at 4C. The
mouse antibody was detected with alkaline-phosphatase-conjugated
antibody (goat anti-mouse IgG, 1:500; Calbiochem). The mem-
branes were then washed and the bands visualised using nitro blue
tetrazolium chloride/5-bromo-4-chloro-3-indolyl phosphate tolui-
dine salt solution for detection of alkaline phosphatase.
Confocal microscopy
Double-label analysis was carried out on NIH3T3 cells adhered to
12 mm glass cover-slips coated with poly-d-lysine (10 g/ml for
1 h). Cells were fixed in 2% paraformaldehyde, 0.15 M sucrose and
0.1% glutaraldehyde for 10 min and were permeabilised by
incubation with 0.1% Triton X-100 for 10 min. The Triton
treatment was omitted in the case of non-permeabilised cells. Prior
to antibody incubation, cells were treated with NH4Cl for 30 min to
minimise the number of reactive aldehyde groups and blocked for
30 min (room temperature) with 5% fetal bovine serum and 1%
BSA in washing buffer. Cells were then incubated with anti-porin
31 HL (1:80), rabbit anti-caveolin-1 (N20, Santa Cruz Biotechnol-
ogy; 1:1,000) or rabbit anti-Tom20 (FL-145, Santa Cruz Biotech-
nology; 1:100) for 2 h at room temperature. Unbound antibody was
removed by washing the cells 3 times with 1 ml of blocking
solution for 10 min each time. Following several washes in
blocking solution, cells were incubated with secondary Alexa Fluor
488 goat anti-rabbit (Molecular Probes; 1:500) and goat anti-mouse
IgG coupled to the fluorochrome Cy3 (Amersham; 1:1,000) or
sheep anti-mouse Texas Red (Amersham; 1:100), for 1 h at room
temperature. Prior to incubation, the secondary antibody was
centrifuged at 13,000 gfor 15 min at 4 C in order to pellet any
precipitated constituents. Negative controls were performed in
which the cells were solely incubated with the secondary antibody.
Digital images were taken with a Leica TCS SP confocal
microscope and analysed with Leica confocal software (Heidelberg,
Germany).
RNA extraction and RT-PCR
RNA extraction and RT-PCR were performed as described
previously [16]. In brief, RNA was isolated from C1300 cells
using the Nucleospin RNA II kit (Macherey-Nagel, Germany),
according to the manufacturer’s instructions. Total RNA (1 g) was
reverse-transcribed to cDNA. We used the following primer pair for
PCR amplification of pl-VDAC [9]: forward 50-TGTGTTCATTC-
TTTCTCGTGC-30and reverse 50-CCAGTGTTCGGCGAGAAT-
GAC-30. PCR products were analysed on a 2% agarose gel
containing a final concentration of 0.5 g/ml ethidium bromide.
Results and discussion
The recent identification of an alternative exon in the
murine VDAC-1 gene provided evidence of a plasma-
lemmal form of VDAC (pl-VDAC) with an N-terminal
leader sequence for its targeting to the plasma membrane
[9]. The presence of a pl-VDAC form in NIH3T3
fibroblasts was confirmed by RT-PCR using primers
specific to the pl-VDAC, including the leader sequence
(Fig. 1A). A band of the expected size (350 bp) was
identified and confirmed to be pl-VDAC by sequencing.
The presence of VDAC protein on NIH3T3 cells was
tested by Western blot (Fig. 1B). The monoclonal anti-
VDAC antibody used (anti-31HL, in particular Ab-2),
was raised against an N-terminal synthetic peptide [17].
Figure 1B shows a Western blot obtained from a
membrane fraction or whole lysate of NIH3T3 cells. A
single band of ~32 kDa, which is the expected size for
VDAC, was obtained from both preparations, indicating
the absence of cross-reactivity with other cellular pro-
teins. Immunolocalisation of VDAC to the plasma
membrane was confirmed by confocal microscopy
(Fig. 2). Non-permeabilised NIH3T3 cells (Fig. 2B) show
membrane delimited staining, consistent with antibody
recognition of the external N-terminal globular a-helix of
the VDAC protein [18]. Permeabilisation of NIH3T3 cells
(Fig. 2E) resulted in staining of the cytosol, most likely
reflecting its organelle localisation, and a reinforcement
of the membrane signal.
Another piece of evidence suggesting the presence of
VDAC in the plasma membrane has been its isolation
from liquid-ordered membrane microdomains called lipid
rafts and its functional reconstitution in artificial bilayers
[8]. To establish whether VDAC was also present in such
specialised membrane microdomains in NIH3T3 cells, we
carried out colocalisation experiments using confocal
microscopy. An antibody against caveolin-1, a protein
present in structured lipid rafts with flask-shaped mem-
brane invaginations termed caveolae [19, 20], was used.
Figure 3 shows confocal images of NIH3T3 cells probed
with anti-VDAC antibody in red (Fig. 3A), and anti-
caveolin-1 antibody in green (Fig. 3B). The merged image
Fig. 1A, B Voltage-dependent anion channel (VDAC) detection in
NIH3T3 cells. AAliquots (10 g) of plasma membrane proteins or
total protein lysate from NIH3T3 fibroblasts were used to detect
VDAC by Western blot analysis. BRT-PCR analysis of pl-VDAC,
which recognises a band of 350 bp that includes the leader
sequence. For the negative control the cDNA was omitted from the
reaction. Western blot and RT-PCR data are representative of at
least 4–10 different experiments
310
(Fig. 3C) shows a marked overlapping of caveolin-1 and
VDAC at the plasma membrane level. These results
reinforced previous observations suggesting the presence
of VDAC at the plasma membrane and, similar to other
cell types [8], VDAC in NIH3T3 fibroblasts also segre-
gates to the caveolin-1-containing lipid rafts.
As mentioned above, eukaryotic VDAC was first
discovered in the outer membrane of mitochondria [1]
where it forms part of the mitochondrial permeability
transition pore (reviewed in [21]). Mitochondria have
always been seen as a homogeneous population of
organelles, the main task of which is the production of
the cell’s energy [21]. Over the last few years, this view
has changed tremendously. Mitochondria contribute to the
shaping and regulation of Ca2+ signals [22] and mito-
chondrial dysfunction leads to different forms of cell
death [21]. Moreover, it seems that, at least in certain cell
types, different pools of mitochondria exist [23]. There-
fore, we evaluated whether VDAC was uniformly
expressed in NIH3T3 mitochondria (Fig. 4). As a marker
of mitochondria we used an anti-Tom20 antibody. Tom20
is an outer mitochondrial membrane protein that forms
part of the translocation machinery involved in the
recognition and translocation of proteins targeted to the
mitochondria. Tom20 has also been proposed as the
receptor protein responsible for the insertion of VDAC in
the outer membrane of the mitochondria [24]. Overlap-
ping of images obtained with anti-VDAC and anti-Tom20
antibodies (Fig. 4C) revealed a perinuclear pool of
mitochondria positive for Tom20 (green) and a peripheral
pool of mitochondria where VDAC and Tom20 colo-
calised (yellow), surrounded by the red signal provided by
the membrane localisation of VDAC. Such differential
localisation of mitochondrial VDAC has not been report-
ed before. We established whether this pattern is unique
for the NIH3T3 fibroblasts or reflects a more general
pattern. The same colocalisation studies were repeated in
another cell line, the C1300 neuroblastoma cell line, that
also expresses maxi-Clchannel activity in the plasma
membrane [25]. Figure 4F shows an even more vivid
tricolour pattern: green (perinuclear mitochondria stained
with Tom20 antibody), yellow (colocalisation of Tom20
and VDAC in peripheral mitochondria) and red (mem-
brane localisation of VDAC). The existence of different
Fig. 3A–C Localisation of VDAC and caveolin-1-containing lipid
rafts in NIH3T3 cells. APermeabilised NIH3T3 cells immuno-
stained with anti-porin 31 HL (VDAC) antibody and Cy3-coupled
secondary antibody (red). BImmunostaining with anti-caveolin-1
antibody and Alexa Fluor 488 secondary antibody (green). C
Merged image of Aand Bshowing extensive colocalisation of
caveolin-1 and VDAC (yellow). Images were obtained by confocal
laser scanning microscopy and are representative of at least 10
different immunolocalisations
Fig. 2A–F Confocal microscopy imaging of VDAC. Non-perme-
abilised (A,Band C) and permeabilised (D,Eand F) NIH3T3 cells
immunostained with anti-porin 31 HL (VDAC) antibody and Cy3-
coupled secondary antibody (red). Controls (Aand D) were
obtained by incubation solely with the secondary antibody. C,F
Transmitted light images of the cells. They are representative of at
least three different immunolocalisations
311
functional pools of mitochondria has been identified in
chromaffin [22] and pancreatic acinar cells [23]. Based on
the differential expression of Tom20 and VDAC proteins
we now demonstrate that in NIH3T3 fibroblasts and
C1300 neuroblastoma cells, at least two pools of mito-
chondria are present.
VDAC appears to be involved in both apoptotic release
of cytochrome c [5] and Ca2+ signalling [26]. The
relevance of this observation is still far from clear. While
there is no evidence suggesting the involvement of a
specific pool of mitochondria in the events leading to
apoptotic cell death, there are several studies reporting the
presence of a peripheral“ring” of mitochondria which
would protect the inner environment from massive
increases in Ca2+ [22, 23]. It would be interesting to
evaluate the precise part played by the two pools of
mitochondria in apoptotic cell death and to establish
whether there is any link between the plasma membrane
VDAC and mitochondrial VDAC in the events leading to
cell apoptosis.
Acknowledgements This work was supported by the Human
Frontiers Science Program, Spanish Ministry of Science and
Technology, Fondo de Investigaciones Sanitarias and Distinci de
la Generalitat de Catalunya per a la Promoci de la Recerca
Universitria. We thank E. Vazquez and J.M. Fernndez-Fernndez
for critical discussion and A. Currid for proof reading the
manuscript.
References
1. Colombini M (1994) Anion channels in the mitochondrial outer
membrane. In Guggino WR (ed.) Chloride channels. Academic
Press, San Diego, pp. 73–101
2. Rostovtseva T, Colombini M (1996) ATP flux is controlled by
a voltage-gated channel from the mitochondrial outer mem-
brane. J Biol Chem 271:28006–28008
3. Papadopoulos V et al. (1997) Peripheral benzodiazepine
receptor in cholesterol transport and steroidogenesis. Steroids
62:21–28
4. Shimizu S, Narita M, Tsujimoto Y (1999) Bcl-2 family proteins
regulate the release of apoptogenic cytochrome c by the
mitochondrial channel VDAC. Nature 399:483–487
5. Shimizu S, Matsuoka Y, Shinohara Y, Yoneda Y, Tsujimoto Y
(2001) Essential role of voltage-dependent anion channel in
various forms of apoptosis in mammalian cells. J Cell Biol
152:237–250
6. Reymann S, et al. (1995) Further evidence for multitopological
localization of mammalian porin (VDAC) in the plasmalemma
forming part of a chloride channel complex affected in cystic
fibrosis and encephalomyopathy. Biochem Mol Med 54:75–87
7. Bathori G, et al. (2000) Extramitochondrial porin: facts and
hypotheses. J Bioenerg Biomembr 32:79–89
8. Bathori G, et al. (1999) Porin is present in the plasma
membrane where it is concentrated in caveolae and caveolae-
related domains. J Biol Chem 274:29607–29612
9. Buettner R, Papoutsoglou G, Scemes E, Spray DC, Dermietzel
R (2000) Evidence for secretory pathway localization of a
voltage-dependent anion channel isoform. Proc Natl Acad Sci
USA 97:3201–3206
10. Blatz AL, Magleby KL (1983) Single voltage-dependent
chloride-selective channels of large conductance in cultured
rat muscle. Biophys J 43:237–241
11. Hardy SP, Valverde MA (1994) Novel plasma membrane
action of estrogen and antiestrogens revealed by their regula-
tion of a large conductance chloride channel. FASEB J. 8:760–
765
12. Valverde MA, Hardy SP, Diaz M (2002) Activation of Maxi
Clchannels by antiestrogens and phenothiazines in NIH3T3
fibroblasts. Steroids 67:439–445
13. Thinnes FP (1992) Evidence for extra-mitochondrial localiza-
tion of the VDAC/porin channel in eukaryotic cells. J Bioenerg
Biomembr 24:71–75
14. Yu WH, Forte M (1996) Is there VDAC in cell compartments
other than the mitochondria? J Bioenerg Biomembr 28:93–100
15. Graeser D, Neubig RR (1992) In Milligan G (ed.) Signal
transduction: a practical approach, IRL Press, Oxford, pp 1–29
16. Fernandez-Fernandez JM, Nobles M, Currid A, Vazquez E,
Valverde MA (2002) Maxi K+channel mediates regulatory
volume decrease response in a human bronchial epithelial cell
line. Am J Physiol 283:C1705–C1714
17. Babel D et al. (1991) Studies on human porin. VI. Production
and characterization of eight monoclonal mouse antibodies
against the human VDAC“Porin 31HL” and their application
for histotopological studies in human skeletal muscle. Biol
Chem Hoppe Seyler 372:1027–1034
18. Casadio R, Jacoboni I, Messina A, De Pinto V (2002) A 3D
model of the voltage-dependent anion channel (VDAC). FEBS
Lett 520:1–7
Fig. 4A–F Localisation of VDAC and Tom20 in NIH3T3 and
C1300 mitochondria. Detection of VDAC and Tom20 in NIH3T3
cells (Aand B, respectively) and C1300 cells (Dand E,
respectively). C,FMerged images. Images were obtained by
confocal laser scanning microscopy and are representative of at
least four different immunolocalisations
312
19. Tsui-Pierchala BA, Encinas M, Milbrandt J, Johnson EM
(2002) Lipid rafts in neuronal signaling and function. Trends
Neurosci. 25:412–417
20. Galbiati F, Razani B, Lisanti MP (2001) Emerging themes in
lipid rafts and caveolae. Cell 106:403–411
21. Krieger C, Duchen MR (2002) Mitochondria, Ca2+ and
neurodegenerative disease. Eur J Pharmacol 447:177–188
22. Villalobos C et al. (2002) Redistribution of Ca2+ among cytosol
and organella during stimulation of bovine chromaffin cells.
FASEB J 16:343–353
23. Park MK, Ashby MC, Erdemli G, Petersen OH, Tepikin AV
(2001) Perinuclear, perigranular and sub-plasmalemmal mito-
chondria have distinct functions in the regulation of cellular
calcium transport. EMBO J 20:1863–1874
24. Schleiff E, Silvius JR, Shore GC (1999) Direct membrane
insertion of voltage-dependent anion-selective channel protein
catalyzed by mitochondrial Tom20. J Cell Biol 145:973–978
25. Diaz M et al. (2001) Okadaic acid-sensitive activation of Maxi
Clchannels by triphenylethylene antioestrogens in C1300
neuroblastoma cells. J Physiol (Lond) 536:79–88
26. Ichas F, Jouaville LS, Mazat JP (1997) Mitochondria are
excitable organelles capable of generating and conveying
electrical and calcium signals. Cell 89:1145–1153
313
... Theoretically, the synaptic mechanism can contribute to neuroprotective properties of olesoxime and represent a new pathway for regulation of neurotransmission by cholesterol-like molecules. Olesoxime targets to voltagedependent anion channels (VDACs) in the outer mitochondrial membrane and this interaction is required for inhibition of mitochondrial permeability transition pore complex, oxidative stress and apoptosis [34][35][36][37][38]. Also, VDACs are localized in the neuronal plasma membrane, particularly in cholesterol-rich microdomains [39][40][41][42][43][44][45]. Functions of plasma membrane VDACs could be linked with control of anion permeability, cell volume, apoptosis, amyloid toxicity, redox status and estrogen receptor α-dependent signaling [39][40][41][42][43][44][46][47][48]. ...
... Olesoxime targets to voltagedependent anion channels (VDACs) in the outer mitochondrial membrane and this interaction is required for inhibition of mitochondrial permeability transition pore complex, oxidative stress and apoptosis [34][35][36][37][38]. Also, VDACs are localized in the neuronal plasma membrane, particularly in cholesterol-rich microdomains [39][40][41][42][43][44][45]. Functions of plasma membrane VDACs could be linked with control of anion permeability, cell volume, apoptosis, amyloid toxicity, redox status and estrogen receptor α-dependent signaling [39][40][41][42][43][44][46][47][48]. Furthermore, VDACs have been detected in proteome of the presynaptic membrane, active zones and SVs [49][50][51][52][53]. ...
... Typically, VDACs represent integral membrane proteins forming pore for small hydrophilic molecules in the outer membrane of mitochondria [73,74]. However, several studies pointed on a localization of VDACs in the plasmalemma [39,[42][43][44][45], including neuronal and synaptic plasma membranes [75][76][77][78]. Consistent with these studies, there was a pattern of VDAC immunolabeling observed in the NMJs. ...
Article
Olesoxime is a cholesterol-like neuroprotective compound that targets to mitochondrial voltage dependent anion channels (VDACs). VDACs were also found in the plasma membrane and highly expressed in the presynaptic compartment. Here, we studied the effects of olesoxime and VDAC inhibitors on neurotransmission in the mouse neuromuscular junction. Electrophysiological analysis revealed that olesoxime suppressed selectively evoked neurotransmitter release in response to a single stimulus and 20 Hz activity. Also olesoxime decreased the rate of FM1–43 dye loss (an indicator of synaptic vesicle exocytosis) at low frequency stimulation and 20 Hz. Furthermore, an increase in extracellular Cl− enhanced the action of olesoxime on the exocytosis and olesoxime increased intracellular Cl− levels. The effects of olesoxime on the evoked synaptic vesicle exocytosis and [Cl−]i were blocked by membrane-permeable and impermeable VDAC inhibitors. Immunofluorescent labeling pointed on the presence of VDACs on the synaptic membranes. Rotenone-induced mitochondrial dysfunction perturbed the exocytotic release of FM1–43 and cell-permeable VDAC inhibitor (but not olesoxime or impermeable VDAC inhibitor) partially mitigated the rotenone-driven alterations in the FM1–43 unloading and mitochondrial superoxide production. Thus, olesoxime restrains neurotransmission by acting on plasmalemmal VDACs whose activation can limit synaptic vesicle exocytosis probably via increasing anion flux into the nerve terminals.
... Immunofluorescence, flow cytometry, and EM immune-gold labelling have detected VDAC in other cell compartments in addition to mitochondria [150,151]. These compartments include the plasma membrane [151,152] (pl-VDAC1) of epithelial cells, astrocytes, and neurons [150,153] as well as in the brain post-synaptic membrane fraction [94]. VDAC1 is localized in isolated caveolae and caveolae-like domains [154], representing microstructures of the plasma membrane. ...
... In VDAC1 heterozygotic mice (VDAC1+/−), mRNA levels of the AD-related genes, including Aβ, APP, and tau, were reduced [175]. Moreover, VDAC1 is required for Aβ cell penetration and cell death induction [149][150][151][152][153][154][155][156][157][158][159][160][162][163][164][165]176]. Furthermore, VDAC1 was proposed to serve as an "amyloid-regulated" channel involved in apoptosis [76,177,178]. ...
... VDAC1 has been found in the plasma membrane [151,155,206] (see Section 8). VDAC1 has been localized in the plasma membrane of neurons [153], and also in isolated caveolae, where it is associated with caveolin-1, a representative scaffolding protein of lipid rafts [76,154,177]. Interestingly, VDAC1 isolated from lipid rafts was found to be associated with γ-secretase [166], suggesting the involvement of VDAC1 in Aβ processing. ...
... It was important that the VDAC channel had a wide nano-sized pore with blockage by and permeability to ATP [98][99][100]. Therefore, it was natural to suppose that this protein somehow reaches the plasma membrane and functions there as the maxi-anion channel [57,84,[101][102][103][104][105][106]. This idea was supported by biochemical isolation of the VDAC protein from preparations of plasma membranes of different origins and by finding of the genetic mechanism of generation of the plasmalemmally expressed VDAC protein (pl-VDAC) through its targeting of the plasma membrane with a signal peptide encoded by an alternative first exon [102]. ...
... The signal peptide is eventually cleaved away, yielding the plasmalemmal VDAC identical to the mitochondrial protein. We have indeed found the presence of mRNA coding for pl-VDAC in the mammary C127 cells (Sabirov and Okada, unpublished observation) confirming results obtained in mouse NIH3T3 fibroblasts [103], C1300 neuroblastoma cells [104], and fibroblasts from wild-type, but not vdac1−/−, mice [57]. However, the Maxi-Cl activity was recorded in fibroblasts isolated from mice with deletion of all individual vdac1, vdac2, and vdac3 genes encoding the three isoforms of the VDAC protein, as well as in cells isolated from vdac1/vdac3 doubledeficient knock-out animals [61]. ...
Article
Full-text available
The Maxi-Cl phenotype accounts for the majority (app. 60%) of reports on the large-conductance maxi-anion channels (MACs) and has been detected in almost every type of cell, including placenta, endothelium, lymphocyte, cardiac myocyte, neuron, and glial cells, and in cells originating from humans to frogs. A unitary conductance of 300–400 pS, linear current-to-voltage relationship, relatively high anion-to-cation selectivity, bell-shaped voltage dependency, and sensitivity to extracellular gadolinium are biophysical and pharmacological hallmarks of the Maxi-Cl channel. Its identification as a complex with SLCO2A1 as a core pore-forming component and two auxiliary regulatory proteins, annexin A2 and S100A10 (p11), explains the activation mechanism as Tyr23 dephosphorylation at ANXA2 in parallel with calcium binding at S100A10. In the resting state, SLCO2A1 functions as a prostaglandin transporter whereas upon activation it turns to an anion channel. As an efficient pathway for chloride, Maxi-Cl is implicated in a number of physiologically and pathophysiologically important processes, such as cell volume regulation, fluid secretion, apoptosis, and charge transfer. Maxi-Cl is permeable for ATP and other small signaling molecules serving as an electrogenic pathway in cell-to-cell signal transduction. Mutations at the SLCO2A1 gene cause inherited bone and gut pathologies and malignancies, signifying the Maxi-Cl channel as a perspective pharmacological target.
... In addition to the mitochondrial membrane, VDAC has also been detected in other cell compartments [16,[55][56][57][58][59][60][61][62][63][64][65], including the plasma membrane [16,57], the sarcoplasmic reticulum (SR) of skeletal muscles [66], and the endoplasmic reticulum (ER) of the rat cerebellum [64,67]. ...
... Interestingly, when detected in the plasma membrane (pl-VDAC1), the amino acid residues that were exposed to the cytosol in the mitochondrial protein were found to face the extracellular space [55,60,[68][69][70][71]. This was demonstrated in epithelial cells, astrocytes, and neurons [56,57] and in differentiated hippocampal neurons [55]. VDAC1 has also been identified in the brain post-synaptic membrane fraction [60] and in the caveolae or caveolae-related domains of established T lymphoid-like cell lines [58]. ...
Article
Full-text available
The voltage-dependent anion channel 1 (VDAC1) protein, is an important regulator of mitochondrial function, and serves as a mitochondrial gatekeeper, with responsibility for cellular fate. In addition to control over energy sources and metabolism, the protein also regulates epigenomic elements and apoptosis via mediating the release of apoptotic proteins from the mitochondria. Apoptotic and pathological conditions, as well as certain viruses, induce cell death by inducing VDAC1 overexpression leading to oligomerization, and the formation of a large channel within the VDAC1 homo-oligomer. This then permits the release of pro-apoptotic proteins from the mitochondria and subsequent apoptosis. Mitochondrial DNA can also be released through this channel, which triggers type-Ι interferon responses. VDAC1 also participates in endoplasmic reticulum (ER)-mitochondria cross-talk, and in the regulation of autophagy, and inflammation. Its location in the outer mitochondrial membrane, makes VDAC1 ideally placed to interact with over 100 proteins, and to orchestrate the interaction of mitochondrial and cellular activities through a number of signaling pathways. Here, we provide insights into the multiple functions of VDAC1 and describe its involvement in several diseases, which demonstrate the potential of this protein as a druggable target in a wide variety of pathologies, including cancer.
... While Kvβ1.1 was distributed evenly, Kvβ2.1 mostly appeared as a punctate pattern. Certain punctate patterns of ion channels indicate protein localization in discrete lipid raft microdomains [26]. Because Kvβ2.1 did not interact with β-actin but was present in the plasma membrane, showing a punctate pattern, whether Kvβ2.1 targeted lipid raft microdomains was postulated (Fig. 2H). ...
Article
Full-text available
The voltage-dependent potassium (Kv) channel Kvβ family was the frst identifed group of modulators of Kv channels. Kvβ regulation of the α-subunits, in addition to their aldoketoreductase activity, has been under extensive study. However, scarce information about their specifc α-subunit-independent biology is available. The expression of Kvβs is ubiquitous and, similar to Kv channels, is tightly regulated in leukocytes. Although Kvβ subunits exhibit cytosolic distribution, spatial localization, in close contact with plasma membrane Kv channels, is crucial for a proper immune response. Therefore, Kvβ2.1 is located near cell surface Kv1.3 channels within the immunological synapse during lymphocyte activation. The objective of this study was to analyze the structural elements that participate in the cellular distribution of Kvβs. It was demonstrated that Kvβ peptides, in addition to the cytoplasmic pattern, targeted the cell surface in the absence of Kv channels. Furthermore, Kvβ2.1, but not Kvβ1.1, targeted lipid raft microdomains in an S-acylation-dependent manner, which was concomitant with peptide localization within the immunological synapse. A pair of C-terminal cysteines (C301/C311) was mostly respon- sible for the specifc palmitoylation of Kvβ2.1. Several insults altered Kvβ2.1 membrane localization. Therefore, growth factor-dependent proliferation enhanced surface targeting, whereas PKC activation impaired lipid raft expression. However, PSD95 stabilized Kvβ2.1 in these domains. This data shed light on the molecular mechanism by which Kvβ2.1 clusters into immunological synapses during leukocyte activation.
... The extra-mitochondrial location was verified by detection of a leader sequence for transport into the plasma membrane [53] and by isolation as part of caveolae [54]. Physiologically it is able to reduce diferric transferrin and releases ferrous iron to be taken up in the cell where it is reoxidized and stored in ferritin [52] [55]. Semidehydroascorbate is another acceptor that is reduced by one-electron reduction to ascorbate, and thus can be recycled as antioxidant [56] [57]. ...
... 3) VDAC has a crucial role in apoptotic mitochondrial changes by interacting with Bax/Bak and Bcl-x L . 4) In neurons, VDAC is also localized in the plasma membrane, 5) especially in isolated caveolae and caveolae-like domains. 6) In Alzheimer's disease (AD) brains, accumulation of VDAC in caveolae is detected in dystrophic neurites of senile plaques. ...
Article
4,4-Diisothiocyanatostilbene disulfonic acid (DIDS), an antagonist of anion channel including voltage-dependent anion channel (VDAC), acts as both neurotoxicant and neuroprotectant, resulting in the controversy. VDAC contributes to neuronal apoptosis and is a candidate target protein of 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2). Caspase-3 is activated during neuronal apoptosis caused by 15d-PGJ2. In the present study, we ascertained whether DIDS was neuroprotective or neurotoxic in the primary culture of rat cortical neurons. Neuronal cell viabilities were primarily evaluated by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H tetrazolium bromide (MTT) reduction assay. Plasma membrane integrity and apoptosis were detected by the staining of propidium iodide (PI) and Hoechst33342, respectively. Alternatively, apoptosis was also measured by caspase-3 assay kit. DIDS did not prevent neurons from undergoing the 15d-PGJ2-induced apoptosis. In contrast, DIDS caused neuronal cell death in a concentration-dependent manner by itself, confirming its neurotoxicity. The sublethal application of DIDS did not decrease MTT-reducing activity, increase caspase-3 activity, condense chromatin, allow PI to enter neuron and degenerate neuronal morphology significantly. Interestingly, DIDS enhanced the 15d-PGJ2-induced neuronal apoptosis markedly under the sublethal condition. To our knowledge, this is the first report of synergistic effects of DIDS on the neurotoxicity of 15d-PGJ2. Graphical Abstract Fullsize Image
... Several host cell proteins that might be the potential targets of EhSSP1 were identified (Fig. 5A). Among these, the most likely candidate was VDAC due to its location on both the plasma membrane and the outer mitochondrial membrane (OMM) (49,50). Also, this would explain a role for EhSSP1 in the interaction of meronts with the PV membrane and in the interaction of the sporoplasm with the plasma cell membrane within the invasion synapse. ...
Article
Full-text available
Microsporidia are important opportunistic human pathogens in immune-suppressed individuals, such as those with HIV/AIDS and recipients of organ transplants. The sporoplasm is critical for establishing microsporidian infection. Despite the biological importance of this structure for transmission, there is limited information about its structure and composition that could be targeted for therapeutic intervention. Here, we identified a novel E. hellem sporoplasm surface protein, EhSSP1, and demonstrated that it can bind to host cell mitochondria via host VDAC. Our data strongly suggest that the interaction between SSP1 and VDAC is important for the association of mitochondria with the parasitophorous vacuole during microsporidian infection. In addition, binding of SSP1 to the host cell is associated with the final steps of invasion in the invasion synapse.
... Conductive release of ATP from cells is associated with 2 types of plasma membrane channels, the Cl À channels, such as maxi-ion channels and volume-regulated ion channels, or the pore forming channels, such as connexins and pannexins. The maxi-ion channels have been found in many cell types, such as endothelial cells (Bahamonde and Valverde, 2003), placental cells (Riquelme, 2009), and in various immune cells (McCann et al., 1989;Kolb and Ubl, 1987;Schlichter et al., 1990). These channels allow passage of small organic anions, such as ATP , and are activated by e.g. ...
Article
Extracellular adenosine 5′-triphosphate (ATP) and adenosine molecules are intimately involved in immune responses. ATP is mostly a pro-inflammatory molecule and is released during hypoxic condition and by necrotic cells, as well as by activated immune cells and endothelial cells. However, under certain conditions, for instance at low concentrations or at prolonged exposure, ATP may also have anti-inflammatory properties. Extracellular ATP can activate both P2X and P2Y purinergic receptors. Extracellular ATP can be hydrolyzed into adenosine in a two-step enzymatic process involving the ectonucleotidases CD39 (ecto-apyrase) and CD73. These enzymes are expressed by many cell types, including endothelial cells and immune cells. The counterpart of ATP is adenosine, which is produced by breakdown of intra- or extracellular ATP. Adenosine has mainly anti-inflammatory effects by binding to the adenosine, or P1, receptors (A1, A2A, A2B, and A3). These receptors are also expressed in many cells, including immune cells. The final effect of ATP and adenosine in immune responses depends on the fine regulatory balance between the 2 molecules. In the present review, we will discuss the current knowledge on the role of these 2 molecules in the immune responses.
Chapter
In addition to their roles in intracellular energy metabolism and nucleic acid synthesis, ATP and other nucleotides play important functions as extracellular signaling molecules (Figure 5.1). It is now more than 30 years since Burnstock1 first proposed that extracellular adenine nucleotides and nucleosides can be used for signal transduction at nerve endings in diverse tissues. Implicit in this notion of purine-based neurotransmission was a requirement for ATP (or other nucleotides) to be released and then degraded in a highly localized fashion at sites of cell-to-cell communication. Given the early emphasis on the role of purines in neuronal signaling, initial studies were focused on nucleotide/nucleoside release at neuron-to-neuron synapses, neuron-to-tissue varicosities, or the immediate vicinity of neuroendocrine cells (e.g., adrenal chromaffin cells).2,3 At approximately the same time, studies from the hematological literature were showing that platelets also release large amounts of ATP and ADP during activation of hemostasis and degranulation of dense granules.4-6 These early investigations demonstrated that neurons, neuroendocrine cells, and platelets could all release ATP via classical mechanisms involving exocytotic release of nucleotides copackaged with biogenic amines or other neurotransmitters within specialized secretory vesicles or granules (Figure 5.1 and Figure 5.2).
Article
Full-text available
Mitochondrial porin, or VDAC, is a pore-forming protein abundant in the outer mitochondrialmembrane. Several publications have reported extramitochondrial localizations as well, butthe evidence was considered insufficient by many, and the presence of porin in nonmitochondrialcellular compartments has remained in doubt for a long time. We have now obtained newdata indicating that the plasma membrane of hematopoietic cells contains porin, probablylocated mostly in caveolae or caveolae-like domains. Porin was purified from the plasmamembrane of intact cells by a procedure utilizing the membrane-impermeable labeling reagentNH-SS-biotin and streptavidin affinity chromatography, and shown to have the same propertiesas mitochondrial porin. A channel with properties similar to that of isolated VDAC wasobserved by patch-clamping intact cells. This review discusses the evidence supportingextramitochondrial localization, the putative identification of the plasma membrane porin with themaxi chloride channel, the hypothetical mechanisms of sorting porin to various cellularmembrane structures, and its possible functions.
Article
Full-text available
The expression of bacterial porin in outer membranes of gram-negative bacteria and of mitochondrial porin or voltage-dependent anion channel (VDAC) in outer mitochondrial membranes (OMM) of eucaryotic cells was demonstrated about 15 years ago. However, the expression of VDAC in the plasmalemma (PLM) of transformed human B lymphoblasts has recently been indicated by cytotoxicity and indirect immunofluorescence studies. New data suggest that the expression of VDAC may be even more widespread. Different cell types express porin channels in their PLM and in intracellular membranes other than OMM. The functional expression of these channels may differ in the various compartments since recent experiments have demonstrated that the voltage dependence and ion selectivity of mitochondrial VDAC may be altered by their interaction with modulators. The present paper proposes a unifying concept for the ion-selective channels of cell membranes, in particular, those whose regulation is affected in cystic fibrosis.
Article
Full-text available
We report on the production and characterization of eight monoclonal mouse antibodies against the complete human VDAC "Porin 31HL". The antigen used was purified from a total membrane preparation of the transformed human B-lymphocyte cell line H2LCL. In Western blots all eight mAbs react with a single 31-kDa band in solubilized H2LCL membrane preparations thus demonstrating their specificity for the human VDAC "Porin 31HL". Concerning the epitope specificity we show that all eight mAbs equally react with the N-terminal part of human porin. Moreover, we demonstrate the expression of VDAC in the sarcolemma by indirect immunoenzyme labelling of cryosections of human skeletal muscle applying four of our mAbs. These data support our recent observations on the expression of porin channels in the plasmalemma of different normal and transformed human cell lines. VDAC in the plasmalemma is discussed as the molecular basis of the Blatz and Magleby channel.
Article
Since the publication of the first edition of Signal Transduction: A Practical Approach in 1992 there has been a great deal of new information about the processes of signal transduction and consequently many new methods have been developed. This new edition has therefore been updated and extended to include the major new methods now available. The first part of the book is mainly concerned with G protein-coupled receptors and covers structural studies of conformational changes and binding sites, phosphorylation and desensitisation, identification, receptor fusion proteins, and reporter gene systems. The second part includes methods for studying components of the other major families of signal transduction: adenylyl cylase and cAMP, phosphorylated inositol lipids, phosphinositide 3-kinases, phosphlipase D and phosphatidylcholine, sphingosine kinase, and inositol 1,4,5-triphosphate. Also included are chapters on baculoviral expression systems and the quantitative assay of mitogen activated protein kinases in intact cells and tissues. As with the previous edition Signal Transduction 2e covers a wide range of techniques and will be useful to both experienced researchers and newcomers.
Article
At the beginning phase, the reconstitution of an anion-selective channel, from mitochondria, came as a surprise. The existence of large channels in the outer membrane, favoring anions, fits nicely with the major function of mitochondria, energy transduction. Substrates and products are mostly negatively charged molecules, such as pyruvate, adenosine diphosphate (ADP), adenosine triphosphate (ATP), phosphate, etc. The reconstitution into planar phospholipid membranes of large voltage gated channels, called VDAC (voltage-dependent anion-selective channel), indicated that these channels are not static structures but dynamic and under regulation. VDAC channels form extremely conductive pathways in phospholipid membranes. Such channels are the primary pathway for the flow of metabolites across the mitochondrial outer membrane. Their well-conserved properties include a variety of regulatory mechanisms that could restrict the flow of metabolites between the cytoplasm and the mitochondrion. Such a bottleneck could limit such things, as energy production and mitochondrial growth and reproduction. Thus, VDAC may play an important role in these and other processes. The importance of VDAC and its properties to mitochondrial function is strongly indicated by the remarkable conservation of its structure and functional properties. Discovery of new regulatory mechanisms and their remarkable conservation provides further evidence for an important and elaborate regulatory system.
Article
• The regulation of Maxi Cl− channels by 17-oestradiol and non-steroidal triphenylethylene antioestrogens represents a rapid, non-classical effect of these compounds. In the present study we have investigated the signalling pathways used for the regulation of Maxi Cl− channel activity by oestrogens and antioestrogens in C1300 neuroblastoma cells. • Whole-cell Maxi Cl− currents were readily and reversibly activated by tamoxifen, toremifene and the membrane-impermeant ethyl-bromide tamoxifen, only when applied to the extracellular medium. • Pre-treatment of C1300 cells with oestrogen or cAMP prevented the antioestrogen-induced activation of Maxi Cl− channels. The inhibitory effect of 17-oestradiol and cAMP was abolished by the kinase inhibitor staurosporine. • Current activation was unaffected by the removal of intracellular Ca2+ and Mg2+, but was completely abolished in the presence of okadaic acid. These results are consistent with the participation of an okadaic acid-sensitive serine/threonine protein phosphatase in the activation of Maxi Cl− channels. However, neither oestrogen or antioestrogen treatment modified the total activity of the two major serine/threonine phosphatases, PP1 and PP2A, in C1300 cells. • Although the role of these Maxi Cl− channels remains unknown, our findings suggest strongly that their modulation by oestrogens and antioestrogens is linked to intracellular signalling pathways.
Article
Single-channel currents of an anion-selective channel in the plasma membrane of cultured rat muscle cells (myotubes) were recorded with the patch-clamp technique (Hamill, O.P., A. Marty, E. Neher, B. Sakmann, and F.J. Sigworth, 1981. Pfluegers Arch. Eur. J. Physiol., 391:85-100). The channel is selective for Cl- over cations, and has an unusually large single-channel conductance of approximately 430 pS in symmetrical 143 mM KCl. The channel is often active at 0 mV, opening and closing spontaneously. When active, steps from 0 mV to either negative or positive membrane potentials close the channel to an apparent inactivated state. The mean effective time that a channel is open before it inactivates is approximately 1.19 s for steps to -30 mV and 0.48 s for steps to +30 mV. Returning the membrane potential to 0 mV results in recovery from inactivation. Calcium ions are not required for channel activity.
Article
Antiestrogens antagonize many genomic effects of estrogen through binding to the nuclear estrogen receptor. We report here that NIH3T3 fibroblasts grown in the presence of colchicine acquire the activation of a large conductance chloride channel upon exposure to extracellular but not intracellular antiestrogens. This effect can be prevented by extracellular 17 beta-estradiol, but not intracellular 17 beta-estradiol or extracellular 17 alpha-estradiol. This is the first demonstration of a regulatory role for antiestrogens and estrogens in the regulation of ionic channels occurring through an interaction of these compounds with a plasma membrane binding site distinct from the classical estrogen receptor and subsequent activation of intracellular second messenger pathway (or pathways).